Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 2(5): e429, 2022 May.
Article in English | MEDLINE | ID: mdl-35598301

ABSTRACT

Human hepatocytes are a critical resource for a broad array of in vitro studies that are conducted during drug development. Cryopreserved human hepatocytes offer great advantages in terms of ease of use and lot-specific repeatability. However, many important cell functions may be diminished or lost altogether in thawed cells. Freshly isolated human hepatocytes may provide the ideal platform for in vitro studies, but limited access to tissue, the challenges of generating high-quality cell preparations, and the innate variability of multiple donors can all be confounding factors. Recently, freshly isolated hepatocytes from chimeric humanized mice have become available (Yamasaki et al., 2010). These PXB-cells feature both the ease of use and lot-specific reproducibility that is provided by cryopreserved human hepatocytes, but are freshly isolated on demand and therefore avoid the complication of diminished cellular function. PXB-cells are prepared at a cell density of 2.1 × 105 cells/cm2 , delivered within 7 days post-seeding in most of the cases, and can maintain the major drug metabolism factors at least up to 21 days post-seeding (Yamasaki et al., 2020). © 2022 Wiley Periodicals LLC. Basic Protocol: Care and culture of PXB-cells Support Protocol: Preparation of dHCGM medium for PXB-cells.


Subject(s)
Hepatocytes , Liver , Animals , Chimera , Mice , Reproducibility of Results
2.
Comp Med ; 72(6): 355-363, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36744513

ABSTRACT

Humanized liver chimeric mice (PXB-mice) are generated by the transplantation of human hepatocytes into mice that have severe combined immunodeficiency and express an albumin-promoted urokinase-type plasminogen activator (cDNA-uPA/SCID) transgene. Human hepatocytes cannot synthesize ascorbic acid (AA; commonly called vitamin C) and humans require supplementation to prevent vitamin C deficiency. PXB-mouse livers contain up to approximately 95% human hepatocytes, which likely affects AA synthesis. To determine whether dietary AA supplementation prevents scurvy-like symptoms and death in PXB-mice, a 12 week study that compared nonsupplemented and supplemented PXB-mice was conducted. Approximately 4 weeks into the study, PXB-mice without dietary supplementation of AA displayed weight loss and clinical signs of hypovitaminosis C, including hunched posture, unkempt appearance, and lameness. Pathologic evaluation of nonsupplemented PXB-mice revealed lesions consistent with hypovitaminosis C. Mean serum AA concentrations in the nonsupplemented PXB-mice were below the limit of quantitation (0.5 µg/mL) and were substantially less than those of controls. AA was also measured in a number of tissues, including adrenal gland, brain, liver, and testis; low AA concentrations were similarly observed in tissues obtained from the nonsupplemented PXB-mice. Collectively, these findings support AA supplementation in PXB-mice to prevent the development of hypovitaminosis C and the potential utility of nonsupplemented PXB-mice as an animal model of scurvy.


Subject(s)
Scurvy , Male , Mice , Humans , Animals , Mice, SCID , Liver , Hepatocytes , Models, Animal
3.
J Hepatol ; 74(5): 1075-1086, 2021 05.
Article in English | MEDLINE | ID: mdl-33333207

ABSTRACT

BACKGROUND & AIMS: While certain nucleos(t)ide reverse transcriptase inhibitors (NRTIs) are efficacious in treating HBV infection, their effects are yet to be optimized and the emergence of NRTI-resistant HBV variants is an issue because of the requirement for lifelong treatment. The development of agents that more profoundly suppress wild-type and drug-resistant HBVs, and that have a long-acting effect, are crucial to improve patient outcomes. METHODS: Herein, we synthesized a novel long-acting 4'-modified NRTI termed E-CFCP. We tested its anti-HBV activity in vitro, before evaluating its anti-HBV activity in HBV-infected human-liver-chimeric mice (PXB-mice). E-CFCP's long-acting features and E-CFCP-triphosphate's interactions with the HBV reverse transcriptase (HBV-RT) were examined. RESULTS: E-CFCP potently blocked HBVWTD1 production (IC50qPCR_cell=1.8 nM) in HepG2.2.15 cells and HBVWTC2 (IC50SB_cell=0.7 nM), entecavir (ETV)-resistant HBVETV-RL180M/S202G/M204V (IC50SB_cell=77.5 nM), and adefovir-resistant HBVADV-RA181T/N236T production (IC50SB_cell=14.1 nM) in Huh7 cells. E-CFCP profoundly inhibited intracellular HBV DNA production to below the detection limit, but ETV and tenofovir alafenamide (TAF) failed to do so. E-CFCP also showed less toxicity than ETV and TAF. E-CFCP better penetrated hepatocytes and was better tri-phosphorylated; E-CFCP-triphosphate persisted intracellularly for longer than ETV-triphosphate. Once-daily peroral E-CFCP administration over 2 weeks (0.02~0.2 mg/kg/day) reduced HBVWTC2-viremia by 2-3 logs in PXB-mice without significant toxicities and the reduction persisted over 1-3 weeks following treatment cessation, suggesting once-weekly dosing capabilities. E-CFCP also reduced HBVETV-RL180M/S202G/M204V-viremia by 2 logs over 2 weeks, while ETV completely failed to reduce HBVETV-RL180M/S202G/M204V-viremia. E-CFCP's 4'-cyano and fluorine interact with both HBVWT-RT and HBVETV-RL180M/S202G-M204 -RT via Van der Waals and polar forces, being important for E-CFCP-triphosphate's interactions and anti-HBV potency. CONCLUSION: E-CFCP represents the first reported potential long-acting NRTI with potent activity against wild-type and treatment-resistant HBV. LAY SUMMARY: Although there are currently effective treatment options for HBV, treatment-resistant variants and the need for lifelong therapy pose a significant challenge. Therefore, the development of new treatment options is crucial to improve outcomes and quality of life. Herein, we report preclinical evidence showing that the anti-HBV agent, E-CFCP, has potent activity against wild-type and treatment-resistant variants. In addition, once-weekly oral dosing may be possible, which is preferrable to the current daily dosing regimens.


Subject(s)
Drug Development/methods , Drug Resistance, Viral/drug effects , Hepatitis B virus , Hepatitis B , Reverse Transcriptase Inhibitors/pharmacology , Animals , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Drug Administration Routes , Drug Administration Schedule , Hepatitis B/drug therapy , Hepatitis B/virology , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Humans , Mice , RNA-Directed DNA Polymerase/metabolism , Time
4.
Xenobiotica ; 50(11): 1370-1379, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32401667

ABSTRACT

We previously reported a prediction method for human pharmacokinetics (PK) using single species allometric scaling (SSS) and the complex Dedrick plot in chimeric mice with humanized liver to predict the total clearance (CLt), distribution volumes in steady state (Vdss) and plasma concentration-time profiles of several drugs metabolized by cytochrome P450 (P450) and non-P450 enzymes. In the present study, we examined eight compounds (bosentan, cerivastatin, fluvastatin, pitavastatin, pravastatin, repaglinide, rosuvastatin, valsartan) as typical organic anion transporting polypeptide (OATP) substrates and six compounds metabolized by P450 and non-P450 enzymes to evaluate the predictability of CLt, Vdss and plasma concentration-time profiles after intravenous administration to chimeric mice. The predicted CLt and Vdss of drugs that undergo OATP-mediated uptake and P450/non-P450-mediated metabolism reflected the observed data from humans within a threefold error range. We also examined the possibility of predicting plasma concentration-time profiles of drugs that undergo OATP-mediated uptake using the complex Dedrick plot in chimeric mice. Most profiles could be superimposed with observed profiles from humans within a two- to threefold error range. PK prediction using SSS and the complex Dedrick plot in chimeric mice can be useful for evaluating drugs that undergo both OATP-mediated uptake and P450/non-P450-mediated metabolism.


Subject(s)
Liver/metabolism , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/metabolism , Animals , Humans , Inactivation, Metabolic , Metabolic Clearance Rate , Mice , Pharmacokinetics
5.
Biomed Res ; 41(1): 33-42, 2020.
Article in English | MEDLINE | ID: mdl-32092738

ABSTRACT

We investigated lipid metabolism in PXB-cells, which are human primary hepatocytes isolated from liver-humanized mice, and HepG2 and HuH-7 human hepatoma cell lines. Lipoprotein levels were higher in PXB-cells than in the 2 other cell lines, and PXB-cells mainly released triglycerides and cholesterol as very low density lipoprotein (VLDL), similar to actual liver tissue, whereas the major lipoprotein released from the 2 hepatoma cell lines was LDL. RT-PCR analysis demonstrated that the gene expression levels of apolipoprotein B100 (ApoB100), the apolipoprotein of VLDL/LDL, were similar in PXB-cells and HepG2 cells, while the overexpression of ApoC2, ApoC3, and ApoE, which are components of VLDL, but not LDL, was observed in PXBcells. A protein immunoassay revealed that ApoB100 levels secreted from PXB-cells and HuH-7 cells were similar; however, ApoC3 levels were higher in PXB-cells than in the two other cell lines. We also examined the anti-lipidemic activities of fenofibrate using this assay system. Fenofibrate suppressed lipoprotein production from PXB-cells in a dose-dependent manner mainly by activating the ß-oxidation pathway. These results suggest that PXB-cells produce high levels of lipoproteins and are suitable for screening anti-lipidemic agents.


Subject(s)
Fenofibrate/pharmacology , Lipid Metabolism , Lipoproteins/metabolism , Animals , Apolipoprotein B-100/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Hep G2 Cells , Hepatocytes , Humans , Lipoproteins, VLDL/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Mice , Mice, SCID , Oxygen/chemistry , Time Factors , Triglycerides/metabolism
6.
J Interferon Cytokine Res ; 39(3): 133-146, 2019 03.
Article in English | MEDLINE | ID: mdl-30844328

ABSTRACT

Interferons (IFNs) suppress viral infection through the induction of >400 interferon-stimulated genes (ISGs). Among ISGs, IFN-induced protein with tetratricopeptide repeats (IFITs) is one of the most potent and well-characterized ISGs. IFIT family consists of 4 cluster genes. It has been suggested that the antiviral action of each IFIT employs distinct mechanisms. In addition, it has been shown that each IFIT exhibits its antiviral properties partially in a pathogen-specific manner. To date, the expression profile of IFITs in the liver, as well as the antiviral potency of the individual IFITs in the regulation of hepatitis C virus (HCV) infection, is not yet fully defined. Our previous study found that the expression of hepatic IFITs is well correlated with the outcome of IFN-based antiviral therapy. This study explored the significance of each IFIT in the suppression of HCV. Our in vitro and in vivo studies with humanized liver chimeric mouse system revealed that IFIT1, 2, and 3/4 play an important role in the suppression of HCV. In addition, our in vitro experiment found that all IFITs possess a comparable anti-HCV potency. Follow-up studies collectively indicated that IFITs suppress HCV likely through 2 distinct mechanisms: (1) inhibition of internal ribosome entry site-dependent viral protein translation initiation complex according to experiments with bicistronic reporter assay as well as confocal microscopic analyses and (2) sequestration of viral genome based on an experiment using replication defective viral genome. In conclusion, our study defined the importance of IFITs in the regulation of HCV and also suggested the multifaceted antiviral actions.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Interferons/pharmacology , Cell Line , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/metabolism , Humans , Interferons/genetics , Tetratricopeptide Repeat , Virus Replication/drug effects , Virus Replication/genetics
7.
Article in English | MEDLINE | ID: mdl-30744813

ABSTRACT

Genotoxicity assays are characterized by a method, an in vitro or in vivo target, and an endpoint. Many cell types have been used as targets, including bacterial cells, cultured mammalian cells, and rodent cells in vivo. Human cells are the most important target for evaluating the risk to humans associated with exposure to chemicals. Almost exclusively, the human cells used in genotoxicity tests have been cultured cells. Here, we have tested human hepatocytes in PXB-mice®, chimeric mice in which the liver has been repopulated with human hepatocytes, as a source of target cells for in vivo genotoxicity assays. We applied the single-cell gel electrophoresis (comet) assay to detect DNA damage and the micronucleus assay to evaluate chromosomal aberrations. These chimeric mice can serve as a valuable model system for genotoxicity assays.


Subject(s)
Chimera/physiology , Chromosome Aberrations , DNA Damage , Hepatocytes/pathology , Mutagenicity Tests/methods , Mutagens/adverse effects , Animals , Comet Assay , Hepatocytes/drug effects , Humans , Mice , Micronucleus Tests
8.
Toxicol Sci ; 167(1): 190-201, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30202993

ABSTRACT

The activation of tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor 2 (TRAIL-R2)/death receptor 5 (DR5) induces apoptosis in various tumor cells but not in normal human cells. Because some therapeutic antibodies targeting TRAIL-R2 have demonstrated severe hepatotoxicity in clinical applications, novel in vivo models reflecting clinical hepatotoxicity are now required. In this study, we investigated the hepatotoxicity caused by KMTR2, an anti-human TRAIL-R2 monoclonal antibody, in chimeric mice with humanized livers (PXB-mice). PXB-mice were exposed to KMTR2 by single or repeated (weekly for 4 weeks) intravenous administrations, and the analyses of blood chemistry, liver histopathology, hepatic gene expression, and toxicokinetics were performed. Treatment with 1 or 10 mg/kg of KMTR2 increased alanine transaminase (ALT) activity and human ALT1 levels in blood. Histopathological analysis revealed that cell death and degeneration with the infiltration of inflammatory cells in human but not mouse hepatocytes were increased in a time-dependent manner after KMTR2 administration. Furthermore, increases in TdT-mediated dUTP nick end labeling (TUNEL)-positive human hepatocytes and serum concentration of cleaved cytokeratin 18, a human-specific apoptosis marker, were observed. RNA sequence analysis showed that the gene expression profile changed in different manners between human and mouse hepatocytes and the up-regulation of TRAIL-R2-related genes was observed only in human hepatocytes. Taken together, these results indicate that KMTR2-mediated TRAIL-R2 activation induces apoptosis of human hepatocytes and hepatotoxicity in PXB-mice and suggest that chimeric mice with humanized liver can be novel tools for the evaluation of in vivo human-specific hepatotoxicity induced by therapeutic antibodies in pre-clinical studies.


Subject(s)
Antibodies, Monoclonal/toxicity , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Animals , Biomarkers/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chimera , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/transplantation , Humans , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Mice , Mice, SCID , Mice, Transgenic
9.
ACS Infect Dis ; 5(5): 738-749, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30408957

ABSTRACT

Hepatitis delta virus (HDV) infects 10-20 million individuals worldwide and causes severe fulminant hepatitis with high likelihood of cirrhosis and hepatocellular carcinoma. HDV infection cannot occur in the absence of the surface antigen (HBsAg) of the hepatitis B virus. RNA interference is an effective mechanism by which to inhibit viral transcripts, and siRNA therapeutics sharing this mechanism have begun to demonstrate clinical efficacy. Here we assessed the outcome of HBV-targeting siRNA intervention against HDV and compared it to a direct anti-HDV siRNA approach in dually infected humanized mice. Treatment with ARB-1740, a clinical stage HBV-targeting siRNA agent delivered using lipid nanoparticle (LNP) technology, effectively reduced HBV viremia by 2.3 log10 and serum HBsAg by 2.6 log10, leading to 1.6 log10 reduction of HDV viremia. In contrast, HDV-targeting siRNA inhibited HDV in both blood and liver compartments without affecting HBV and PEGylated interferon-alpha reduced HBV viremia by 2.0 log10 but had no effect on HDV viremia under these study conditions. These results illustrate the inhibitory effects of siRNAs against these two viral infections and suggest that ARB-1740 may be of therapeutic benefit for hepatitis delta patients, a subpopulation with high unmet medical need.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis D/drug therapy , Hepatitis Delta Virus/drug effects , RNA Interference , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice
10.
Drug Metab Dispos ; 46(1): 11-19, 2018 01.
Article in English | MEDLINE | ID: mdl-29051147

ABSTRACT

The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and DDIs via transporters may be a risk factor for adverse events. Cyclosporine A, a strong OATP inhibitor, has been reported to increase the systemic exposure of rosuvastatin, an OATP substrate, by 7.1-fold in clinical studies. PXB mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug discovery in drug metabolism and pharmacokinetics studies. In the present study, we examined in vivo and in vitro DDIs between rosuvastatin and cyclosporine A in PXB mice and fresh human hepatocytes (PXB cells) obtained from PXB mice. We initially investigated the active transport of rosuvastatin into PXB cells, and found concentration-dependent uptake with a Michaelis-Menten constant value of 4.0 µmol/l and a Vmax value of 4.63 pmol/min per 106 cells. Cyclosporine A inhibited the uptake of rosuvastatin with an IC50 value of 0.21 µmol/l. We then examined in vivo DDIs, and the exposure of orally administered rosuvastatin increased by 3.3-fold and 11-fold in PXB mice pretreated with 10 and 50 mg/kg cyclosporine A, whereas it increased by 2.5-fold and 6.2-fold when rosuvastatin was administered intravenously, in studies that were conducted for considering gastrointestinal DDIs. The liver-to-blood concentration ratio of rosuvastatin was dose-dependently decreased by pretreatment with cyclosporine A in PXB mice and SCID mice. Observed DDIs in vivo were considered to be reasonable based on the estimated concentrations of cyclosporine A at the inlet to the liver and in the liver tissues of both mice. In conclusion, our results indicate that PXB mice might be a useful tool for predicting human OATP-mediated DDIs in drug discovery, and its limitation due to the differences of gastrointestinal condition from human should also be considered.


Subject(s)
Cyclosporine/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Organic Anion Transporters/metabolism , Rosuvastatin Calcium/pharmacology , Transplantation Chimera/metabolism , Animals , Drug Interactions , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Liver/cytology , Liver/metabolism , Male , Mice , Mice, SCID , Models, Animal , Organic Anion Transporters/antagonists & inhibitors
11.
J Hepatol ; 64(3): 547-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26505121

ABSTRACT

BACKGROUND & AIMS: Antiviral agents including entecavir (ETV) suppress the replication of the hepatitis B virus (HBV) genome in human hepatocytes, but they do not reduce the abundance of viral proteins. The present study focused on effectively reducing viral protein levels. METHODS: We designed siRNAs (HBV-siRNA) that target consensus sequences in HBV genomes. To prevent the emergence of escaped mutant virus, we mixed three HBV-siRNAs (HBV-siRNAmix); the mixture was encapsulated in a novel pH-sensitive multifunctional envelope-type nanodevice (MEND), a hepatocyte-specific drug delivery system. Coagulation factor 7 siRNA was used to assess delivery and knockdown efficiencies of MEND/siRNA treatments in mice. The potency of MEND/HBV-siRNAmix was evaluated in primary human hepatocytes and in chimeric mice with humanized liver persistently infected with HBV. RESULTS: Effective knockdown of targets, efficient delivery of siRNA, and liver-specific delivery were each observed with MEND. MEND/HBV-siRNA caused efficient reduction of HBsAg and HBeAg in vitro and in vivo. However, ETV treatment did not efficiently reduce HBsAg or HBeAg when compared with a single MEND/HBV-siRNAmix treatment. Furthermore, the suppressive effects of a single dose of MEND/HBV-siRNAmix persisted for 14days in vitro and in vivo. CONCLUSION: We demonstrated that MEND/HBV-siRNA controlled HBV more efficiently than did ETV. Furthermore, the effect of a single dose of MEND/HBV-siRNA persisted for a long time. These results indicated that MEND/HBV-siRNA may be a promising novel HBV treatment that is more effective than reverse transcriptase inhibitors.


Subject(s)
Gene Transfer Techniques , Hepatitis B, Chronic/therapy , RNA, Small Interfering/administration & dosage , Animals , DNA, Viral/analysis , Hepatitis B Surface Antigens/analysis , Hepatitis B e Antigens/analysis , Hepatitis B virus/genetics , Humans , Hydrogen-Ion Concentration , Liposomes , Mice
12.
Biopharm Drug Dispos ; 37(1): 3-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26352195

ABSTRACT

Chimeric mice with humanized livers (PXB mice) are used to investigate the metabolism and pharmacokinetics of drugs in humans. However, residual murine enzymatic activities derived from the liver and the presence of mouse small intestinal metabolism can hamper the prediction of human drug metabolism. Recently murine Cytochrome P450 3a gene knockout chimeric mice with humanized livers (Cyp3a KO CM) were developed. To evaluate the prediction of drug metabolism, nefazodone (NEF) was administered orally at 10 mg/kg to the following mouse strains: Cyp3a KO CM, murine Cyp3a gene knockout (Cyp3a KO), PXB and severe combined immunodeficiency (SCID) mice. Liquid chromatography-mass spectrometry was used for metabolic profiling of plasma, urine and bile. The prediction of human metabolite levels such as hydroxy nefazodone (OH-NEF), triazoledione form (TD), m-chlorophenylpiperazine and dealkyl metabolites in Cyp3a KO CM was superior to that in Cyp3a KO, PXB or SCID mice. Further, clinical exposure levels of NEF, OH-NEF and TD were reproduced in Cyp3a KO CM. In contrast, NEF was rapidly metabolized to TD in both PXB and SCID mice but not in Cyp3a KO mice, suggesting that murine CYP3A is involved in the elimination of NEF in these mice. These findings demonstrate that the metabolic profile of NEF in Cyp3a KO CM differs qualitatively and quantitatively from that in PXB mice due to the higher metabolic rate of NEF and its metabolites via murine CYP3A. Therefore Cyp3a KO CM might be useful in predicting the metabolic profiles of drug candidates in humans.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Liver/metabolism , Triazoles/pharmacokinetics , Animals , Antidepressive Agents, Second-Generation/blood , Antidepressive Agents, Second-Generation/urine , Bile/chemistry , Child, Preschool , Cytochrome P-450 CYP3A/genetics , Hepatocytes/metabolism , Humans , Male , Mice, Inbred ICR , Mice, Knockout , Mice, SCID , Microsomes, Liver/metabolism , Piperazines , Triazoles/blood , Triazoles/urine
13.
PLoS One ; 10(11): e0142145, 2015.
Article in English | MEDLINE | ID: mdl-26536627

ABSTRACT

We have used homozygous albumin enhancer/promoter-driven urokinase-type plasminogen activator/severe combined immunodeficient (uPA/SCID) mice as hosts for chimeric mice with humanized livers. However, uPA/SCID mice show four disadvantages: the human hepatocytes (h-heps) replacement index in mouse liver is decreased due to deletion of uPA transgene by homologous recombination, kidney disorders are likely to develop, body size is small, and hemizygotes cannot be used as hosts as more frequent homologous recombination than homozygotes. To solve these disadvantages, we have established a novel host strain that has a transgene containing albumin promoter/enhancer and urokinase-type plasminogen activator cDNA and has a SCID background (cDNA-uPA/SCID). We applied the embryonic stem cell technique to simultaneously generate a number of transgenic lines, and found the line with the most appropriate levels of uPA expression-not detrimental but with a sufficiently damaged liver. We transplanted h-heps into homozygous and hemizygous cDNA-uPA/SCID mice via the spleen, and monitored their human albumin (h-alb) levels and body weight. Blood h-alb levels and body weight gradually increased in the hemizygous cDNA-uPA/SCID mice and were maintained until they were approximately 30 weeks old. By contrast, blood h-alb levels and body weight in uPA/SCID chimeric mice decreased from 16 weeks of age onwards. A similar decrease in body weight was observed in the homozygous cDNA-uPA/SCID genotype, but h-alb levels were maintained until they were approximately 30 weeks old. Microarray analyses revealed identical h-heps gene expression profiles in homozygous and hemizygous cDNA-uPA/SCID mice were identical to that observed in the uPA/SCID mice. Furthermore, like uPA/SCID chimeric mice, homozygous and hemizygous cDNA-uPA/SCID chimeric mice were successfully infected with hepatitis B virus and C virus. These results indicate that hemizygous cDNA-uPA/SCID mice may be novel and useful hosts for producing chimeric mice for use in future long-term studies, including hepatitis virus infection analysis or drug toxicity studies.


Subject(s)
Chimerism , Disease Models, Animal , Hepatitis, Viral, Human , Liver/metabolism , Mice, Inbred Strains/genetics , Animals , Breeding , Child , Child, Preschool , Female , Hemizygote , Hepatitis Viruses/pathogenicity , Hepatocytes/transplantation , Humans , Liver/cytology , Male , Mice, Inbred Strains/virology , Mice, SCID
15.
Drug Metab Dispos ; 42(6): 1055-65, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700822

ABSTRACT

Human-specific or disproportionately abundant human metabolites of drug candidates that are not adequately formed and qualified in preclinical safety assessment species pose an important drug development challenge. Furthermore, the overall metabolic profile of drug candidates in humans is an important determinant of their drug-drug interaction susceptibility. These risks can be effectively assessed and/or mitigated if human metabolic profile of the drug candidate could reliably be determined in early development. However, currently available in vitro human models (e.g., liver microsomes, hepatocytes) are often inadequate in this regard. Furthermore, the conduct of definitive radiolabeled human ADME studies is an expensive and time-consuming endeavor that is more suited for later in development when the risk of failure has been reduced. We evaluated a recently developed chimeric mouse model with humanized liver on uPA/SCID background for its ability to predict human disposition of four model drugs (lamotrigine, diclofenac, MRK-A, and propafenone) that are known to exhibit human-specific metabolism. The results from these studies demonstrate that chimeric mice were able to reproduce the human-specific metabolite profile for lamotrigine, diclofenac, and MRK-A. In the case of propafenone, however, the human-specific metabolism was not detected as a predominant pathway, and the metabolite profiles in native and humanized mice were similar; this was attributed to the presence of residual highly active propafenone-metabolizing mouse enzymes in chimeric mice. Overall, the data indicate that the chimeric mice with humanized liver have the potential to be a useful tool for the prediction of human-specific metabolism of xenobiotics and warrant further investigation.


Subject(s)
Chimera/genetics , Chimera/metabolism , Hepatocytes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Hepatocytes/drug effects , Humans , Liver/drug effects , Liver/metabolism , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Mice , Mice, SCID , Mice, Transgenic , Pharmaceutical Preparations/administration & dosage
16.
Int J Mol Sci ; 15(1): 58-74, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24362577

ABSTRACT

We performed in vivo and in vitro studies to determine the induction of human cytochrome P450 (CYP) using chimeric mice with humanized liver (PXB-mice®) and human hepatocytes isolated from the PXB-mice (PXB-cells), which were derived from the same donor. For the in vivo study, PXB-mice were injected with 3-methylcholanthrene (3-MC, 2 or 20 mg/kg) or rifampicin (0.1 or 10 mg/kg) for four days. For the in vitro study, PXB-cells were incubated with 3-MC (10, 50, or 250 ng/mL) or with rifampicin (5 or 25 µg/mL). The CYP1A1 and 1A2, and CYP3A4 mRNA expression levels increased significantly in the PXB-mouse livers with 20 mg/kg of 3-MC (Cmax, 12.2 ng/mL), and 10 mg/kg rifampicin (Cmax, 6.9 µg/mL), respectively. The CYP1A1 mRNA expression level increased significantly in PXB-cells with 250 ng/mL of 3-MC, indicating lower sensitivity than in vivo. The CYP1A2 and CYP3A4 mRNA expression levels increased significantly with 50 ng/mL of 3-MC, and 5 µg/mL of rifampicin, respectively, which indicated that the sensitivities were similar between in vivo and in vitro studies. In conclusion, PXB-mice and PXB-cells provide a robust model as an intermediate between in vivo and in vitro human metabolic enzyme induction studies.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP3A/biosynthesis , Hepatocytes/enzymology , Liver/enzymology , Animals , Chimera , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP3A/genetics , Enzyme Induction , Half-Life , Humans , Immunohistochemistry , Keratin-18/metabolism , Keratin-8/metabolism , Methylcholanthrene/pharmacokinetics , Mice , RNA, Messenger/metabolism , Rifampin/pharmacokinetics
17.
Gut ; 62(9): 1340-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23135762

ABSTRACT

OBJECTIVE: Recent studies have demonstrated that genetic polymorphisms near the IL28B gene are associated with the clinical outcome of pegylated interferon α (peg-IFN-α) plus ribavirin therapy for patients with chronic hepatitis C virus (HCV). However, it is unclear whether genetic variations near the IL28B gene influence hepatic interferon (IFN)-stimulated gene (ISG) induction or cellular immune responses, lead to the viral reduction during IFN treatment. DESIGN: Changes in HCV-RNA levels before therapy, at day 1 and weeks 1, 2, 4, 8 and 12 after administering peg-IFN-α plus ribavirin were measured in 54 patients infected with HCV genotype 1. Furthermore, we prepared four lines of chimeric mice having four different lots of human hepatocytes containing various single nucleotide polymorphisms (SNP) around the IL28B gene. HCV infecting chimeric mice were subcutaneously administered with peg-IFN-α for 2 weeks. RESULTS: There were significant differences in the reduction of HCV-RNA levels after peg-IFN-α plus ribavirin therapy based on the IL28B SNP rs8099917 between TT (favourable) and TG/GG (unfavourable) genotypes in patients; the first-phase viral decline slope per day and second-phase slope per week in TT genotype were significantly higher than in TG/GG genotype. On peg-IFN-α administration to chimeric mice, however, no significant difference in the median reduction of HCV-RNA levels and the induction of antiviral ISG was observed between favourable and unfavourable human hepatocyte genotypes. CONCLUSIONS: As chimeric mice have the characteristic of immunodeficiency, the response to peg-IFN-α associated with the variation in IL28B alleles in chronic HCV patients would be composed of the intact immune system.


Subject(s)
Hepacivirus/drug effects , Hepatitis C, Chronic , Interferon-alpha/pharmacology , Interleukins/genetics , Polyethylene Glycols/pharmacology , RNA, Viral/analysis , Ribavirin/pharmacology , Animals , Antiviral Agents/pharmacology , Drug Therapy, Combination , Female , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Interferons , Male , Mice , Mice, SCID , Middle Aged , Polymorphism, Single Nucleotide , Recombinant Proteins/pharmacology , Time Factors , Treatment Outcome , Viral Load/methods
18.
Lab Invest ; 93(1): 54-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23147226

ABSTRACT

We previously produced mice with human hepatocyte (h-hep) chimeric livers by transplanting h-heps into albumin enhancer/promoter-driven urokinase-type plasminogen activator-transgenic severe combined immunodeficient (SCID) mice with liver disease. The chimeric livers were constructed with h-heps, mouse hepatocytes, and mouse hepatic sinusoidal cells (m-HSCs). Here, we investigated the morphological features of the chimeric livers and the h-hep gene expression profiles in the xenogeneic animal body. To do so, we performed immunohistochemistry, morphometric analyses, and electron microscopic observations on chimeric mouse livers, and used microarray analyses to compare gene expression patterns in hepatocytes derived from chimeric mouse hepatocytes (c-heps) and h-heps. Morphometric analysis revealed that the ratio of hepatocytes to m-HSCs in the chimeric mouse livers were twofold higher than those in the SCID mouse livers, corresponding to twin-cell plates in the chimeric mouse liver. The h-heps in the chimeric mouse did not show hypoxia even in the twin-cell plate structure, probably because of low oxygen consumption by the h-heps relative to the mouse hepatocytes (m-heps). Immunohistochemical and electron microscopic examinations revealed that the sinusoids in the chimeric mouse livers were normally constructed with h-heps and m-HSCs. However, a number of microvilli projected into the intercellular clefts on the lateral aspects of the hepatocytes, features typical of a growth phase. Microarray profiles indicated that ∼82% of 16 605 probes were within a twofold range difference between h-heps and c-heps. Cluster and principal component analyses showed that the gene expression patterns of c-heps were extremely similar to those of h-heps. In conclusion, the chimeric mouse livers were normally reconstructed with h-heps and m-HSCs, and expressed most human genes at levels similar to those in human livers, although the chimeric livers showed morphological characteristics typical of growth.


Subject(s)
Hepatocytes/cytology , Liver/cytology , Analysis of Variance , Animals , Cell Adhesion/physiology , Cell Hypoxia/physiology , Female , Gene Expression Profiling , Hepatic Stellate Cells/cytology , Humans , Immunohistochemistry , Kupffer Cells/cytology , Liver/chemistry , Male , Mice , Mice, SCID , Tissue Array Analysis/methods , Transplantation, Heterologous
19.
Toxicol Lett ; 214(1): 9-18, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22902350

ABSTRACT

Troglitazone (Tro) is a thiazolidinedione antidiabetic drug that was withdrawn from the market due to its association with idiosyncratic severe liver injury. Tro has never induced liver injury in experimental animals in vivo. It was assumed that the species differences between human and experimental animals in the pharmaco- or toxicokinetics of Tro might be associated with these observations. In this study, we investigated whether a chimeric mouse with a humanized liver that we previously established, whose replacement index with human hepatocytes is up to 92% can reproduce Tro-induced liver injury. When the chimeric mice were orally administered Tro for 14 or 23 days (1000mg/kg/day), serum alanine aminotransferase (ALT) was significantly increased by 2.1- and 3.6-fold, respectively. Co-administration of l-buthionine sulfoximine (10mM in drinking water), an inhibitor of glutathione (GSH) synthesis, unexpectedly prevented the Tro-dependent increase of ALT, which suggests that the GSH scavenging pathway will not be involved in Tro-induced liver injury. To elucidate the mechanism of the onset of liver injury, hepatic GSH content, the level of oxidative stress markers and phase I and phase II drug metabolizing enzymes were determined. However, these factors were not associated with Tro-induced liver injury. An immune-mediated reaction may be associated with Tro-induced liver toxicity in vivo, because the chimeric mouse is derived from an immunodeficient SCID mouse. In conclusion, we successfully reproduced Tro-induced liver injury using chimeric mice with a humanized liver, which provides a new animal model for studying idiosyncratic drug-induced liver injury.


Subject(s)
Antineoplastic Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Chromans/toxicity , Thiazolidinediones/toxicity , Transplantation Chimera/metabolism , Animals , Buthionine Sulfoximine/pharmacology , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Liver/metabolism , Liver/pathology , Mice , Troglitazone
20.
Drug Metab Dispos ; 40(3): 474-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22126990

ABSTRACT

The induction of cytochrome P450 (P450) enzymes is one of the risk factors for drug-drug interactions (DDIs). To date, the human pregnane X receptor (PXR)-mediated CYP3A4 induction has been well studied. In addition to CYP3A4, the expression of CYP2C subfamily is also regulated by PXR, and the DDIs caused by the induction of CYP2C enzymes have been reported to have a major clinical impact. The purpose of the present study was to investigate whether chimeric mice with a humanized liver (PXB mice) can be a suitable animal model for investigating the PXR-mediated induction of CYP2C subfamily, together with CYP3A4. We evaluated the inductive effect of rifampicin (RIF), a typical human PXR ligand, on the plasma exposure to the four P450 substrate drugs (triazolam/CYP3A4, pioglitazone/CYP2C8, (S)-warfarin/CYP2C9, and (S)-(-)-mephenytoin/CYP2C19) by cassette dosing in PXB mice. The induction of several drug-metabolizing enzymes and transporters in the liver was also examined by measuring the enzyme activity and mRNA expression levels. Significant reductions in the exposure to triazolam, pioglitazone, and (S)-(-)-mephenytoin, but not to (S)-warfarin, were observed. In contrast to the in vivo results, all the four P450 isoforms, including CYP2C9, were elevated by RIF treatment. The discrepancy in the (S)-warfarin results between in vivo and in vitro studies may be attributed to the relatively small contribution of CYP2C9 to (S)-warfarin elimination in the PXB mice used in this study. In summary, PXB mice are a useful animal model to examine DDIs caused by PXR-mediated induction of CYP2C and CYP3A4.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Receptors, Steroid/metabolism , Animals , Biological Transport , Biotransformation , Chimera , Drug Interactions , Humans , Inactivation, Metabolic , Liver/enzymology , Male , Mephenytoin/metabolism , Mephenytoin/pharmacokinetics , Mice , Microsomes, Liver/metabolism , Models, Animal , Pioglitazone , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rifampin/pharmacology , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacokinetics , Triazolam/metabolism , Triazolam/pharmacokinetics , Warfarin/metabolism , Warfarin/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...